1
|
Morita M, Fujii R, Ryuno A, Morimoto M, Inoko A, Inoue T, Ikenouchi J, Atsuta Y, Hayashi Y, Teramoto T, Saito D. The yolk sac vasculature in early avian embryo provides a novel model for the analysis of cancer extravasation. Dev Biol 2025:S0012-1606(25)00133-2. [PMID: 40381710 DOI: 10.1016/j.ydbio.2025.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 04/21/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
Hematogenous metastasis, a hallmark of cancer cells, involves a complex series of migration steps, including intravasation, circulation, arrest in blood vessels, and trans-endothelial migration (TEM)-the lattar two collectively referred to as extravasation. Among these steps, extravasation poses significant challenges for imaging in amniotes such as humans and mice due to its unpredictable timing and location, which limits our understanding of the underlying cellular and molecular mechanisms. Thus, the development of a novel cancer carrier model with high-resolution imaging capabilities in amniotes is essential. In this study, we investigated the yolk sac vasculature (YSV) of early avian embryos (chickens and quail) as an innovative model for studying extravasation, capitalizing on its superior imaging capabilities. We assessed the YSV structure and applied fluorescent labeling to improve visibility. Following this, cancer cells were introduced into the YSV, and their behavior was monitored, revealing distinct morphologies and dynamics associated with extravasation. Furthermore, the YSV model exhibited a high degree of quantitative precision for extravasation studies and demonstrated potential for drug screening applications. Our findings indicate that the YSV model holds promise as a novel platform for elucidating the cellular and molecular mechanisms involved in cancer metastasis through advanced imaging techniques.
Collapse
Affiliation(s)
- Mizuki Morita
- Graduate School of Systems Life Sciences, Kyushu University
| | - Ryo Fujii
- Graduate School of Systems Life Sciences, Kyushu University
| | - Asuka Ryuno
- Graduate School of Systems Life Sciences, Kyushu University
| | | | - Akihito Inoko
- Department of Pathology, Aichi Medical University School of Medicine, Nagakute, Aichi, 480-1195, Japan; Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Aichi, 464-8681, Japan
| | - Takahiro Inoue
- Department of Nephro-Urologic Surgery and Andrology, Graduate School of Medicine, Mie University, Tsu, Mie, 514-8507, Japan
| | - Junichi Ikenouchi
- Graduate School of Systems Life Sciences, Kyushu University; Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Fukuoka, 819-0395, Japan
| | - Yuji Atsuta
- Graduate School of Systems Life Sciences, Kyushu University; Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Fukuoka, 819-0395, Japan
| | - Yoshiki Hayashi
- Graduate School of Systems Life Sciences, Kyushu University; Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Fukuoka, 819-0395, Japan
| | - Takayuki Teramoto
- Graduate School of Systems Life Sciences, Kyushu University; Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Fukuoka, 819-0395, Japan
| | - Daisuke Saito
- Graduate School of Systems Life Sciences, Kyushu University; Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Fukuoka, 819-0395, Japan.
| |
Collapse
|
2
|
Testa AM, Vignozzi L, Corallo D, Aveic S, Viola A, Allegra M, Angioni R. Hypoxic Human Microglia Promote Angiogenesis Through Extracellular Vesicle Release. Int J Mol Sci 2024; 25:12508. [PMID: 39684220 DOI: 10.3390/ijms252312508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Microglia, the brain-resident immune cells, orchestrate neuroinflammatory responses and are crucial in the progression of neurological diseases, including ischemic stroke (IS), which accounts for approximately 85% of all strokes worldwide. Initially deemed detrimental, microglial activation has been shown to perform protective functions in the ischemic brain. Besides their effects on neurons, microglia play a role in promoting post-ischemic angiogenesis, a pivotal step for restoring oxygen and nutrient supply. However, the molecular mechanisms underlying microglia-endothelial cell interactions remain largely unresolved, particularly in humans. Using both in vitro and in vivo models, we investigated the angiogenic signature and properties of extracellular vesicles (EVs) released by human microglia upon hypoxia-reperfusion stimulation. EVs were isolated and characterized in terms of their size, concentration, and protein content. Their angiogenic potential was evaluated using endothelial cell assays and a zebrafish xenograft model. The in vivo effects were further assessed in a mouse model of ischemic stroke. Our findings identified key proteins orchestrating the pro-angiogenic functions of human microglial EVs under hypoxic conditions. In vitro assays demonstrated that hypoxic EVs (hypEVs) promoted endothelial cell migration and tube formation. In vivo, hypEVs induced vessel sprouting in zebrafish and increased microvessel density in the perilesional area of mice following ischemic stroke.
Collapse
Affiliation(s)
- Alessandra Maria Testa
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
- Laboratory of Immunity, Inflammation and Angiogenesis, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padua, Italy
| | - Livia Vignozzi
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Diana Corallo
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padua, Italy
| | - Sanja Aveic
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padua, Italy
| | - Antonella Viola
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Manuela Allegra
- Laboratory of Neuronal Circuits in Developmental Disorders, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padua, Italy
- Neuroscience Institute, National Research Council, 35131 Padua, Italy
| | - Roberta Angioni
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
- Laboratory of Immunity, Inflammation and Angiogenesis, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padua, Italy
| |
Collapse
|
3
|
Ucci A, Giacchi L, Rucci N. Primary Bone Tumors and Breast Cancer-Induced Bone Metastases: In Vivo Animal Models and New Alternative Approaches. Biomedicines 2024; 12:2451. [PMID: 39595017 PMCID: PMC11591690 DOI: 10.3390/biomedicines12112451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
Bone is the preferential site of metastasis for the most common tumors, including breast cancer. On the other hand, osteosarcoma is the primary bone cancer that most commonly occurs and causes bone cancer-related deaths in children. Several treatment strategies have been developed so far, with little or no efficacy for patient survival and with the development of side effects. Therefore, there is an urgent need to develop more effective therapies for bone primary tumors and bone metastatic disease. This almost necessarily requires the use of in vivo animal models that better mimic human pathology and at the same time follow the ethical principles for the humane use of animal testing. In this review we aim to illustrate the main and more suitable in vivo strategies employed to model bone metastases and osteosarcoma. We will also take a look at the recent technologies implemented for a partial replacement of animal testing.
Collapse
Affiliation(s)
| | | | - Nadia Rucci
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (A.U.); (L.G.)
| |
Collapse
|
4
|
Zhan T, Song W, Jing G, Yuan Y, Kang N, Zhang Q. Zebrafish live imaging: a strong weapon in anticancer drug discovery and development. Clin Transl Oncol 2024; 26:1807-1835. [PMID: 38514602 DOI: 10.1007/s12094-024-03406-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 02/07/2024] [Indexed: 03/23/2024]
Abstract
Developing anticancer drugs is a complex and time-consuming process. The inability of current laboratory models to reflect important aspects of the tumor in vivo limits anticancer medication research. Zebrafish is a rapid, semi-automated in vivo screening platform that enables the use of non-invasive imaging methods to monitor morphology, survival, developmental status, response to drugs, locomotion, or other behaviors. Zebrafish models are widely used in drug discovery and development for anticancer drugs, especially in conjunction with live imaging techniques. Herein, we concentrated on the use of zebrafish live imaging in anticancer therapeutic research, including drug screening, efficacy assessment, toxicity assessment, and mechanism studies. Zebrafish live imaging techniques have been used in numerous studies, but this is the first time that these techniques have been comprehensively summarized and compared side by side. Finally, we discuss the hypothesis of Zebrafish Composite Model, which may provide future directions for zebrafish imaging in the field of cancer research.
Collapse
Affiliation(s)
- Tiancheng Zhan
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Rd, Jinghai District, Tianjin, 301617, People's Republic of China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Wanqian Song
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Rd, Jinghai District, Tianjin, 301617, People's Republic of China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Guo Jing
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Rd, Jinghai District, Tianjin, 301617, People's Republic of China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Yongkang Yuan
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Rd, Jinghai District, Tianjin, 301617, People's Republic of China
| | - Ning Kang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Rd, Jinghai District, Tianjin, 301617, People's Republic of China.
| | - Qiang Zhang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Rd, Jinghai District, Tianjin, 301617, People's Republic of China.
| |
Collapse
|
5
|
Verma SK, Nandi A, Sinha A, Patel P, Mohanty S, Jha E, Jena S, Kumari P, Ghosh A, Jerman I, Chouhan RS, Dutt A, Samal SK, Mishra YK, Varma RS, Panda PK, Kaushik NK, Singh D, Suar M. The posterity of Zebrafish in paradigm of in vivo molecular toxicological profiling. Biomed Pharmacother 2024; 171:116160. [PMID: 38237351 DOI: 10.1016/j.biopha.2024.116160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/05/2024] [Accepted: 01/11/2024] [Indexed: 02/08/2024] Open
Abstract
The aggrandised advancement in utility of advanced day-to-day materials and nanomaterials has raised serious concern on their biocompatibility with human and other biotic members. In last few decades, understanding of toxicity of these materials has been given the centre stage of research using many in vitro and in vivo models. Zebrafish (Danio rerio), a freshwater fish and a member of the minnow family has garnered much attention due to its distinct features, which make it an important and frequently used animal model in various fields of embryology and toxicological studies. Given that fertilization and development of zebrafish eggs take place externally, they serve as an excellent model organism for studying early developmental stages. Moreover, zebrafish possess a comparable genetic composition to humans and share almost 70% of their genes with mammals. This particular model organism has become increasingly popular, especially for developmental research. Moreover, it serves as a link between in vitro studies and in vivo analysis in mammals. It is an appealing choice for vertebrate research, when employing high-throughput methods, due to their small size, swift development, and relatively affordable laboratory setup. This small vertebrate has enhanced comprehension of pathobiology and drug toxicity. This review emphasizes on the recent developments in toxicity screening and assays, and the new insights gained about the toxicity of drugs through these assays. Specifically, the cardio, neural, and, hepatic toxicology studies inferred by applications of nanoparticles have been highlighted.
Collapse
Affiliation(s)
- Suresh K Verma
- School of Biotechnology, KIIT University, Bhubaneswar, India.
| | - Aditya Nandi
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Adrija Sinha
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Paritosh Patel
- School of Biotechnology, KIIT University, Bhubaneswar, India; Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897, Seoul, South Korea
| | | | - Ealisha Jha
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Snehasmita Jena
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Puja Kumari
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno 61137, Czech Republic
| | - Aishee Ghosh
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Ivan Jerman
- National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Raghuraj Singh Chouhan
- Department of Environmental Sciences, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia
| | - Ateet Dutt
- Instituto de Investigaciones en Materiales, UNAM, CDMX, Mexico
| | - Shailesh Kumar Samal
- Unit of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Yogendra Kumar Mishra
- Mads Clausen Institute, NanoSYD, University of Southern Denmark, Alsion 2, Sønderborg DK-6400, Denmark
| | - Rajender S Varma
- Institute for Nanomaterials, Advanced Technologies and Innovation (CxI), Technical University of Liberec (TUL), Studentská 1402/2, Liberec 1 461 17, Czech Republic
| | - Pritam Kumar Panda
- Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897, Seoul, South Korea.
| | - Deobrat Singh
- Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden.
| | - Mrutyunjay Suar
- School of Biotechnology, KIIT University, Bhubaneswar, India.
| |
Collapse
|
6
|
Ding B, Chen Q, Wu Z, Li X, Ding Y, Wu Q, Han L, Wu H. In Vitro and In Vivo Analyses Reveal Tumor-Derived Exosome miR-558 Promotes Angiogenesis in Tongue Squamous Cell Carcinoma by Targeting Heparinase. Technol Cancer Res Treat 2024; 23:15330338241261615. [PMID: 38887096 PMCID: PMC11185026 DOI: 10.1177/15330338241261615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 06/20/2024] Open
Abstract
This study aimed to investigate the role of miR-558 in tumor angiogenesis by targeting heparinase (HPSE) in tongue squamous cell carcinoma (TSCC)-derived exosomes. In the present study, the role of exosome miR-558 in angiogenesis in vitro and in vivo was investigated by cell proliferation, migration, tube formation, subcutaneous tumor formation in mice, and in vivo Matrigel plug assay. The target genes of miR-558 were detected by means of dual luciferase assay. It was found that TSCC cells secrete miR-558 into the extracellular environment, with exosome as the carrier. Human umbilical vein endothelial cells (HUVEC) ingested exosomes, which not only increased the expression level of miR-558, but also enhanced their proliferation, migration, and tube formation functions. In vivo Matrigel plug assay demonstrated that TSCC cell-derived exosome miR-558 promoted neovascularization in vivo. Compared with negative control cells, TSCC cells overexpressing miR-558 formed subcutaneous tumors in nude mice, with larger volume, heavier mass, and more vascularization. Dual luciferase assay confirmed that HPSE was the direct target gene regulated by miR-558. HPSE promoted the proliferation, migration, and tube formation of HUVECs, and the knockout of HPSE could downregulate the pro-angiogenic effect of miR-558. In summary, miR-558 in TSCC exosomes promotes the proliferation, migration, and tube formation of HUVECs by targeting HPSE, and enhancing tumor angiogenesis.
Collapse
Affiliation(s)
- Bixiao Ding
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Nantong University, Nantong, China
| | - Qingwen Chen
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Zhen Wu
- Department of Otolaryngology-Head and Neck Surgery, Changshu Second People's Hospital, Suzhou, China
| | - Xiaoguang Li
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine; Shanghai Key Lab, Shanghai, China
| | - Yuancheng Ding
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Nantong University, Nantong, China
| | - Qiong Wu
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Nantong University, Nantong, China
| | - Liang Han
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Hao Wu
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
7
|
Sun B, Ji WD, Wang WC, Chen L, Ma JY, Tang EJ, Lin MB, Zhang XF. Circulating tumor cells participate in the formation of microvascular invasion and impact on clinical outcomes in hepatocellular carcinoma. Front Genet 2023; 14:1265866. [PMID: 38028589 PMCID: PMC10652898 DOI: 10.3389/fgene.2023.1265866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a common malignant tumor worldwide. Although the treatment strategies have been improved in recent years, the long-term prognosis of HCC is far from satisfactory mainly due to high postoperative recurrence and metastasis rate. Vascular tumor thrombus, including microvascular invasion (MVI) and portal vein tumor thrombus (PVTT), affects the outcome of hepatectomy and liver transplantation. If vascular invasion could be found preoperatively, especially the risk of MVI, more reasonable surgical selection will be chosen to reduce the risk of postoperative recurrence and metastasis. However, there is a lack of reliable prediction methods, and the formation mechanism of MVI/PVTT is still unclear. At present, there is no study to explore the possibility of tumor thrombus formation from a single circulating tumor cell (CTC) of HCC, nor any related study to describe the possible leading role and molecular mechanism of HCC CTCs as an important component of MVI/PVTT. In this study, we review the current understanding of MVI and possible mechanisms, discuss the function of CTCs in the formation of MVI and interaction with immune cells in the circulation. In conclusion, we discuss implications for potential therapeutic targets and the prospect of clinical treatment of HCC.
Collapse
Affiliation(s)
- Bin Sun
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wei-Dan Ji
- Department of Molecular Oncology, Eastern Hepatobiliary Surgical Hospital and National Center for Liver Cancer, Navy Military Medical University, Shanghai, China
| | - Wen-Chao Wang
- Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lei Chen
- Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jun-Yong Ma
- Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai, China
| | - Er-Jiang Tang
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Mou-Bin Lin
- Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiao-Feng Zhang
- Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai, China
| |
Collapse
|
8
|
Hiraki HL, Matera DL, Wang WY, Prabhu ES, Zhang Z, Midekssa F, Argento AE, Buschhaus JM, Humphries BA, Luker GD, Pena-Francesch A, Baker BM. Fiber density and matrix stiffness modulate distinct cell migration modes in a 3D stroma mimetic composite hydrogel. Acta Biomater 2023; 163:378-391. [PMID: 36179980 PMCID: PMC10043045 DOI: 10.1016/j.actbio.2022.09.043] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 09/12/2022] [Accepted: 09/16/2022] [Indexed: 01/26/2023]
Abstract
The peritumoral stroma is a complex 3D tissue that provides cells with myriad biophysical and biochemical cues. Histologic observations suggest that during metastatic spread of carcinomas, these cues influence transformed epithelial cells, prompting a diversity of migration modes spanning single cell and multicellular phenotypes. Purported consequences of these variations in tumor escape strategies include differential metastatic capability and therapy resistance. Therefore, understanding how cues from the peritumoral stromal microenvironment regulate migration mode has both prognostic and therapeutic value. Here, we utilize a synthetic stromal mimetic in which matrix fiber density and bulk hydrogel mechanics can be orthogonally tuned to investigate the contribution of these two key matrix attributes on MCF10A migration mode phenotypes, epithelial-mesenchymal transition (EMT), and invasive potential. We develop an automated computational image analysis framework to extract migratory phenotypes from fluorescent images and determine 3D migration metrics relevant to metastatic spread. Using this analysis, we find that matrix fiber density and bulk hydrogel mechanics distinctly contribute to a variety of MCF10A migration modes including amoeboid, single mesenchymal, clusters, and strands. We identify combinations of physical and soluble cues that induce a variety of migration modes originating from the same MCF10A spheroid and use these settings to examine a functional consequence of migration mode -resistance to apoptosis. We find that cells migrating as strands are more resistant to staurosporine-induced apoptosis than either disconnected clusters or individual invading cells. Improved models of the peritumoral stromal microenvironment and understanding of the relationships between matrix attributes and cell migration mode can aid ongoing efforts to identify effective cancer therapeutics that address cell plasticity-based therapy resistances. STATEMENT OF SIGNIFICANCE: Stromal extracellular matrix structure dictates both cell homeostasis and activation towards migratory phenotypes. However decoupling the effects of myriad biophysical cues has been difficult to achieve. Here, we encapsulate electrospun fiber segments within an amorphous hydrogel to create a fiber-reinforced hydrogel composite in which fiber density and hydrogel stiffness can be orthogonally tuned. Quantification of 3D cell migration reveal these two parameters uniquely contribute to a diversity of migration phenotypes spanning amoeboid, single mesenchymal, multicellular cluster, and collective strand. By tuning biophysical and biochemical cues to elicit heterogeneous migration phenotypes, we find that collective strands best resist apoptosis. This work establishes a composite approach to modulate fibrous topography and bulk hydrogel mechanics and identified biomaterial parameters to direct distinct 3D cell migration phenotypes.
Collapse
Affiliation(s)
- Harrison L Hiraki
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Daniel L Matera
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States
| | - William Y Wang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Eashan S Prabhu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Zane Zhang
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI, 481095, United States
| | - Firaol Midekssa
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Anna E Argento
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Johanna M Buschhaus
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States; Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, 48109, United States; Department of Radiology, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Brock A Humphries
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States; Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, 48109, United States; Department of Radiology, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Gary D Luker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States; Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, 48109, United States; Department of Radiology, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Abdon Pena-Francesch
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI, 481095, United States
| | - Brendon M Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, United States.
| |
Collapse
|
9
|
Singhal SS, Garg R, Mohanty A, Garg P, Ramisetty SK, Mirzapoiazova T, Soldi R, Sharma S, Kulkarni P, Salgia R. Recent Advancement in Breast Cancer Research: Insights from Model Organisms-Mouse Models to Zebrafish. Cancers (Basel) 2023; 15:cancers15112961. [PMID: 37296923 DOI: 10.3390/cancers15112961] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Animal models have been utilized for decades to investigate the causes of human diseases and provide platforms for testing novel therapies. Indeed, breakthrough advances in genetically engineered mouse (GEM) models and xenograft transplantation technologies have dramatically benefited in elucidating the mechanisms underlying the pathogenesis of multiple diseases, including cancer. The currently available GEM models have been employed to assess specific genetic changes that underlay many features of carcinogenesis, including variations in tumor cell proliferation, apoptosis, invasion, metastasis, angiogenesis, and drug resistance. In addition, mice models render it easier to locate tumor biomarkers for the recognition, prognosis, and surveillance of cancer progression and recurrence. Furthermore, the patient-derived xenograft (PDX) model, which involves the direct surgical transfer of fresh human tumor samples to immunodeficient mice, has contributed significantly to advancing the field of drug discovery and therapeutics. Here, we provide a synopsis of mouse and zebrafish models used in cancer research as well as an interdisciplinary 'Team Medicine' approach that has not only accelerated our understanding of varied aspects of carcinogenesis but has also been instrumental in developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Sharad S Singhal
- Department of Medical Oncology and Therapeutic Research, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Rachana Garg
- Department of Surgery, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Atish Mohanty
- Department of Medical Oncology and Therapeutic Research, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Pankaj Garg
- Department of Chemistry, GLA University, Mathura 281406, Uttar Pradesh, India
| | - Sravani Keerthi Ramisetty
- Department of Medical Oncology and Therapeutic Research, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Tamara Mirzapoiazova
- Department of Medical Oncology and Therapeutic Research, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Raffaella Soldi
- Translational Genomics Research Institute, Phoenix, AZ 85338, USA
| | - Sunil Sharma
- Translational Genomics Research Institute, Phoenix, AZ 85338, USA
| | - Prakash Kulkarni
- Department of Medical Oncology and Therapeutic Research, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
- Department of Systems Biology, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutic Research, Beckman Research Institute, City of Hope Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
10
|
Shakiba D, Genin GM, Zustiak SP. Mechanobiology of cancer cell responsiveness to chemotherapy and immunotherapy: Mechanistic insights and biomaterial platforms. Adv Drug Deliv Rev 2023; 196:114771. [PMID: 36889646 PMCID: PMC10133187 DOI: 10.1016/j.addr.2023.114771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/17/2022] [Accepted: 03/03/2023] [Indexed: 03/08/2023]
Abstract
Mechanical forces are central to how cancer treatments such as chemotherapeutics and immunotherapies interact with cells and tissues. At the simplest level, electrostatic forces underlie the binding events that are critical to therapeutic function. However, a growing body of literature points to mechanical factors that also affect whether a drug or an immune cell can reach a target, and to interactions between a cell and its environment affecting therapeutic efficacy. These factors affect cell processes ranging from cytoskeletal and extracellular matrix remodeling to transduction of signals by the nucleus to metastasis of cells. This review presents and critiques the state of the art of our understanding of how mechanobiology impacts drug and immunotherapy resistance and responsiveness, and of the in vitro systems that have been of value in the discovery of these effects.
Collapse
Affiliation(s)
- Delaram Shakiba
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University, St. Louis, MO, USA; Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, MO, USA
| | - Guy M Genin
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University, St. Louis, MO, USA; Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, MO, USA.
| | - Silviya P Zustiak
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University, St. Louis, MO, USA; Department of Biomedical Engineering, School of Science and Engineering, Saint Louis University, St. Louis, MO, USA.
| |
Collapse
|
11
|
Al-Hamaly MA, Turner LT, Rivera-Martinez A, Rodriguez A, Blackburn JS. Zebrafish Cancer Avatars: A Translational Platform for Analyzing Tumor Heterogeneity and Predicting Patient Outcomes. Int J Mol Sci 2023; 24:2288. [PMID: 36768609 PMCID: PMC9916713 DOI: 10.3390/ijms24032288] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
The increasing number of available anti-cancer drugs presents a challenge for oncologists, who must choose the most effective treatment for the patient. Precision cancer medicine relies on matching a drug with a tumor's molecular profile to optimize the therapeutic benefit. However, current precision medicine approaches do not fully account for intra-tumoral heterogeneity. Different mutation profiles and cell behaviors within a single heterogeneous tumor can significantly impact therapy response and patient outcomes. Patient-derived avatar models recapitulate a patient's tumor in an animal or dish and provide the means to functionally assess heterogeneity's impact on drug response. Mouse xenograft and organoid avatars are well-established, but the time required to generate these models is not practical for clinical decision-making. Zebrafish are emerging as a time-efficient and cost-effective cancer avatar model. In this review, we highlight recent developments in zebrafish cancer avatar models and discuss the unique features of zebrafish that make them ideal for the interrogation of cancer heterogeneity and as part of precision cancer medicine pipelines.
Collapse
Affiliation(s)
- Majd A. Al-Hamaly
- Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40356, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Logan T. Turner
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
- Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40356, USA
| | | | - Analiz Rodriguez
- Department of Neurosurgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jessica S. Blackburn
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
- Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40356, USA
| |
Collapse
|
12
|
Li X, Li M. The application of zebrafish patient-derived xenograft tumor models in the development of antitumor agents. Med Res Rev 2023; 43:212-236. [PMID: 36029178 DOI: 10.1002/med.21924] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/09/2022] [Accepted: 07/28/2022] [Indexed: 02/04/2023]
Abstract
The cost of antitumor drug development is enormous, yet the clinical outcomes are less than satisfactory. Therefore, it is of great importance to develop effective drug screening methods that enable accurate, rapid, and high-throughput discovery of lead compounds in the process of preclinical antitumor drug research. An effective solution is to use the patient-derived xenograft (PDX) tumor animal models, which are applicable for the elucidation of tumor pathogenesis and the preclinical testing of novel antitumor compounds. As a promising screening model organism, zebrafish has been widely applied in the construction of the PDX tumor model and the discovery of antineoplastic agents. Herein, we systematically survey the recent cutting-edge advances in zebrafish PDX models (zPDX) for studies of pathogenesis mechanisms and drug screening. In addition, the techniques used in the construction of zPDX are summarized. The advantages and limitations of the zPDX are also discussed in detail. Finally, the prospects of zPDX in drug discovery, translational medicine, and clinical precision medicine treatment are well presented.
Collapse
Affiliation(s)
- Xiang Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Minyong Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
13
|
Lichtenegger A, Baumann B, Yasuno Y. Optical Coherence Tomography Is a Promising Tool for Zebrafish-Based Research-A Review. Bioengineering (Basel) 2022; 10:5. [PMID: 36671577 PMCID: PMC9854701 DOI: 10.3390/bioengineering10010005] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
The zebrafish is an established vertebrae model in the field of biomedical research. With its small size, rapid maturation time and semi-transparency at early development stages, it has proven to be an important animal model, especially for high-throughput studies. Three-dimensional, high-resolution, non-destructive and label-free imaging techniques are perfectly suited to investigate these animals over various development stages. Optical coherence tomography (OCT) is an interferometric-based optical imaging technique that has revolutionized the diagnostic possibilities in the field of ophthalmology and has proven to be a powerful tool for many microscopic applications. Recently, OCT found its way into state-of-the-art zebrafish-based research. This review article gives an overview and a discussion of the relevant literature and an outlook for this emerging field.
Collapse
Affiliation(s)
- Antonia Lichtenegger
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, 1090 Vienna, Austria
- Computational Optics Group, University of Tsukuba, Tsukuba 305-8573, Japan
| | - Bernhard Baumann
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, 1090 Vienna, Austria
| | - Yoshiaki Yasuno
- Computational Optics Group, University of Tsukuba, Tsukuba 305-8573, Japan
| |
Collapse
|
14
|
Anti-Inflammatory and Antioxidant Properties of Physalis alkekengi L. Extracts In Vitro and In Vivo: Potential Application for Skin Care. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7579572. [PMID: 36310614 PMCID: PMC9605834 DOI: 10.1155/2022/7579572] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/22/2022] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Inflammatory skin disorders are becoming major issues threatening public health with increasing prevalence. This study was to evaluate the anti-inflammatory, antioxidant, and antisenescent activities of traditional folk medicinal plant, Physalis alkekengi L. extracts to alleviate skin inflammation and its possible mechanisms. METHODS Lipopolysaccharides (LPS)-treated murine macrophages RAW264.7 and human skin keratinocytes HaCaT were incubated with the plant extracts, respectively. The production of nitric oxide (NO) was tested by using Griess reagents. The activity of nitric oxide synthase (NOS) was detected through a fluorescence microplate reader. Reactive oxygen species (ROS) production and cell apoptosis were quantified by flow cytometry. The proinflammatory cytokines were measured using ELISA and qRT-PCR. Human skin fibroblasts (HFF-1) were coincubated with D-galactose (D-gal) and the plant extracts. The senescence associated-galactosidase (SA-β-gal) was stained to evaluate cellular senescence. The senescence-associated secretory phenotype (SASP), IL-1β, was measured through ELISA. The mRNA of IL-1α in SLS-stimulated and PGE2 in UV-radiated 3D skin models were detected by qRT-PCR. In vivo ROS production and neutrophil recruitment in CuSO4-treated zebrafish models were observed by fluorescence microscopy. Inflammation-related factors were measured by qRT-PCR. Results. In vitro, Physalis alkekengi L. significantly reduced NO production, NOS activity, cell apoptosis, transcription of TNF-α, IL-6, IL-1β and ROS production. These plant extracts markedly attenuated SA-β-gal and IL-1β and downregulated the production of IL-1α and PGE2. In vivo, the plant extracts dramatically dampened ROS production, the number of neutrophils, and proinflammatory cytokines. CONCLUSIONS Cumulatively, this work systematically demonstrated the anti-inflammatory, antioxidant, and antisenescent properties of Physalis alkekengi L. and proposed the possible roles of Physalis alkekengi L. in inflammatory signaling pathways, providing an effective natural product for the treatment of inflammatory skin disorders.
Collapse
|
15
|
Groenendyk J, Stoletov K, Paskevicius T, Li W, Dai N, Pujol M, Busaan E, Ng HH, Boukouris AE, Saleme B, Haromy A, Cui K, Hu M, Yan Y, Zhang R, Michelakis E, Chen XZ, Lewis JD, Tang J, Agellon LB, Michalak M. Loss of the fructose transporter SLC2A5 inhibits cancer cell migration. Front Cell Dev Biol 2022; 10:896297. [PMID: 36268513 PMCID: PMC9578049 DOI: 10.3389/fcell.2022.896297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 09/09/2022] [Indexed: 11/13/2022] Open
Abstract
Metastasis is the primary cause of cancer patient death and the elevation of SLC2A5 gene expression is often observed in metastatic cancer cells. Here we evaluated the importance of SLC2A5 in cancer cell motility by silencing its gene. We discovered that CRISPR/Cas9-mediated inactivation of the SLC2A5 gene inhibited cancer cell proliferation and migration in vitro as well as metastases in vivo in several animal models. Moreover, SLC2A5-attenuated cancer cells exhibited dramatic alterations in mitochondrial architecture and localization, uncovering the importance of SLC2A5 in directing mitochondrial function for cancer cell motility and migration. The direct association of increased abundance of SLC2A5 in cancer cells with metastatic risk in several types of cancers identifies SLC2A5 as an important therapeutic target to reduce or prevent cancer metastasis.
Collapse
Affiliation(s)
- Jody Groenendyk
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | | | | | - Wenjuan Li
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Ning Dai
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Myriam Pujol
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Erin Busaan
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Hoi Hei Ng
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | | | - Bruno Saleme
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Alois Haromy
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Kaisa Cui
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Miao Hu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, China
| | - Yanan Yan
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, China
| | - Rui Zhang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, China
| | | | - Xing-Zhen Chen
- Department of Physiology, University of Alberta, Edmonton, AB, Canada
| | - John D. Lewis
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Jingfeng Tang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, China
| | - Luis B. Agellon
- School of Human Nutrition, McGill University, Montreal, QC, Canada
- *Correspondence: Luis B. Agellon, ; Marek Michalak,
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Luis B. Agellon, ; Marek Michalak,
| |
Collapse
|
16
|
Lichtenegger A, Tamaoki J, Licandro R, Mori T, Mukherjee P, Bian L, Greutter L, Makita S, Wöhrer A, Matsusaka S, Kobayashi M, Baumann B, Yasuno Y. Longitudinal investigation of a xenograft tumor zebrafish model using polarization-sensitive optical coherence tomography. Sci Rep 2022; 12:15381. [PMID: 36100620 PMCID: PMC9470556 DOI: 10.1038/s41598-022-19483-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/30/2022] [Indexed: 01/19/2023] Open
Abstract
Breast cancer is a leading cause of death in female patients worldwide. Further research is needed to get a deeper insight into the mechanisms involved in the development of this devastating disease and to find new therapy strategies. The zebrafish is an established animal model, especially in the field of oncology, which has shown to be a promising candidate for pre-clinical research and precision-based medicine. To investigate cancer growth in vivo in zebrafish, one approach is to explore xenograft tumor models. In this article, we present the investigation of a juvenile xenograft zebrafish model using a Jones matrix optical coherence tomography (JM-OCT) prototype. Immunosuppressed wild-type fish at 1-month post-fertilization were injected with human breast cancer cells and control animals with phosphate buffered saline in the tail musculature. In a longitudinal study, the scatter, polarization, and vasculature changes over time were investigated and quantified in control versus tumor injected animals. A significant decrease in birefringence and an increase in scattering signal was detected in tumor injected zebrafish in comparison to the control once. This work shows the potential of JM-OCT as a non-invasive, label-free, three-dimensional, high-resolution, and tissue-specific imaging tool in pre-clinical cancer research based on juvenile zebrafish models.
Collapse
Affiliation(s)
- Antonia Lichtenegger
- Computational Optics Group, University of Tsukuba, Tsukuba, Japan.
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria.
| | - Junya Tamaoki
- Department of Molecular and Developmental Biology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Roxane Licandro
- Computational Imaging Research Lab, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
- Laboratory for Computational Neuroimaging, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Tomoko Mori
- Clinical Research and Regional Innovation, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | | | - Lixuan Bian
- Department of Molecular and Developmental Biology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Lisa Greutter
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Shuichi Makita
- Computational Optics Group, University of Tsukuba, Tsukuba, Japan
| | - Adelheid Wöhrer
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Satoshi Matsusaka
- Clinical Research and Regional Innovation, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Makoto Kobayashi
- Department of Molecular and Developmental Biology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Bernhard Baumann
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Yoshiaki Yasuno
- Computational Optics Group, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
17
|
Bera K, Kiepas A, Zhang Y, Sun SX, Konstantopoulos K. The interplay between physical cues and mechanosensitive ion channels in cancer metastasis. Front Cell Dev Biol 2022; 10:954099. [PMID: 36158191 PMCID: PMC9490090 DOI: 10.3389/fcell.2022.954099] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Physical cues have emerged as critical influencers of cell function during physiological processes, like development and organogenesis, and throughout pathological abnormalities, including cancer progression and fibrosis. While ion channels have been implicated in maintaining cellular homeostasis, their cell surface localization often places them among the first few molecules to sense external cues. Mechanosensitive ion channels (MICs) are especially important transducers of physical stimuli into biochemical signals. In this review, we describe how physical cues in the tumor microenvironment are sensed by MICs and contribute to cancer metastasis. First, we highlight mechanical perturbations, by both solid and fluid surroundings typically found in the tumor microenvironment and during critical stages of cancer cell dissemination from the primary tumor. Next, we describe how Piezo1/2 and transient receptor potential (TRP) channels respond to these physical cues to regulate cancer cell behavior during different stages of metastasis. We conclude by proposing alternative mechanisms of MIC activation that work in tandem with cytoskeletal components and other ion channels to bestow cells with the capacity to sense, respond and navigate through the surrounding microenvironment. Collectively, this review provides a perspective for devising treatment strategies against cancer by targeting MICs that sense aberrant physical characteristics during metastasis, the most lethal aspect of cancer.
Collapse
Affiliation(s)
- Kaustav Bera
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
| | - Alexander Kiepas
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
- *Correspondence: Alexander Kiepas, ; Konstantinos Konstantopoulos,
| | - Yuqi Zhang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
| | - Sean X. Sun
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Department of Mechanical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Department of Oncology, The Johns Hopkins University, Baltimore, MD, United States
- *Correspondence: Alexander Kiepas, ; Konstantinos Konstantopoulos,
| |
Collapse
|
18
|
Dudziak K, Nowak M, Sozoniuk M. One Host-Multiple Applications: Zebrafish ( Danio rerio) as Promising Model for Studying Human Cancers and Pathogenic Diseases. Int J Mol Sci 2022; 23:10255. [PMID: 36142160 PMCID: PMC9499349 DOI: 10.3390/ijms231810255] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/03/2022] [Accepted: 09/03/2022] [Indexed: 11/17/2022] Open
Abstract
In recent years, zebrafish (ZF) has been increasingly applied as a model in human disease studies, with a particular focus on cancer. A number of advantages make it an attractive alternative for mice widely used so far. Due to the many advantages of zebrafish, modifications can be based on different mechanisms and the induction of human disease can take different forms depending on the research goal. Genetic manipulation, tumor transplantation, or injection of the pathogen are only a few examples of using ZF as a model. Most of the studies are conducted in order to understand the disease mechanism, monitor disease progression, test new or alternative therapies, and select the best treatment. The transplantation of cancer cells derived from patients enables the development of personalized medicine. To better mimic a patient's body environment, immune-deficient models (SCID) have been developed. A lower immune response is mostly generated by genetic manipulation but also by irradiation or dexamethasone treatment. For many studies, using SCID provides a better chance to avoid cancer cell rejection. In this review, we describe the main directions of using ZF in research, explain why and how zebrafish can be used as a model, what kind of limitations will be met and how to overcome them. We collected recent achievements in this field, indicating promising perspectives for the future.
Collapse
Affiliation(s)
- Karolina Dudziak
- Chair and Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Michał Nowak
- Institute of Plant Genetics, Breeding and Biotechnology, University of Life Sciences in Lublin, 20-950 Lublin, Poland
| | - Magdalena Sozoniuk
- Institute of Plant Genetics, Breeding and Biotechnology, University of Life Sciences in Lublin, 20-950 Lublin, Poland
| |
Collapse
|
19
|
Wang X, Li W, Jiang H, Ma C, Huang M, Wei X, Wang W, Jing L. Zebrafish Xenograft Model for Studying Pancreatic Cancer-Instructed Innate Immune Microenvironment. Int J Mol Sci 2022; 23:6442. [PMID: 35742884 PMCID: PMC9224329 DOI: 10.3390/ijms23126442] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 12/10/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has up to half the tumor mass of tumor-associated myeloid cells. Myeloid innate immune cells play important roles in regulating cancer cell recognition and tumor growth. PDAC cells often mold myeloid cells into pro-tumoral state to fuel cancer growth and induce immune suppression. However, how tumor cells educate the innate immune responses remains largely unknown. In this study, we used four different human PDAC cell lines (PANC1, BxPC3, AsPC1, and CFPAC1) to establish the zebrafish xenograft model and investigated the interaction between pancreatic cancer and innate immune cells. The primary tumor-derived cancer cells PANC1 and BxPC3 activated innate immune anti-tumoral responses efficiently, while cancer cells from metastatic tissues AsPC1 and CFPAC1 induced an innate immune suppression and educated innate immune cells towards pro-tumoral state. Chemical conversion of innate immune cells to anti-tumoral state inhibited tumor growth for AsPC1 and CFPAC1. Moreover, genetic and pharmacological inhibition of macrophages also significantly reduced tumor growth, supporting the important roles of macrophages in innate immune suppression. REG4 expression is high in AsPC1 and CFPAC1. Knockdown of REG4 induced innate immune activation and reduced tumor growth in the xenografts, indicating that REG4 is a beneficial target for PDAC therapy. Our study provides a fast in-vivo model to study PDAC-innate immune interaction and their plasticity that could be used to study the related mechanism as well as identify new drugs to enhance immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lili Jing
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; (X.W.); (W.L.); (H.J.); (C.M.); (M.H.); (X.W.); (W.W.)
| |
Collapse
|
20
|
Qian C, Yu X, Tong M, Zhuang S, Lin W. Visual-Guided Solutions in Automated Zebrafish Larva Heart Micro-Injection. IEEE Robot Autom Lett 2022. [DOI: 10.1109/lra.2021.3140059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Cheng Qian
- Research Institute of Intelligent Control and Systems, Harbin Institute of Technology, Harbin, China
| | - Xinghu Yu
- Ningbo Institute of Intelligent Equipment Technology Co. Ltd., Ningbo, China
| | - Mingsi Tong
- Research Institute of Intelligent Control and Systems, Harbin Institute of Technology, Harbin, China
| | - Songlin Zhuang
- Department of Mechanical Engineering, University of Victoria, Victoria, Canada
| | - Weiyang Lin
- Research Institute of Intelligent Control and Systems, Harbin Institute of Technology, Harbin, China
| |
Collapse
|
21
|
Research Techniques Made Simple: Zebrafish Models for Human Dermatologic Disease. J Invest Dermatol 2022; 142:499-506.e1. [DOI: 10.1016/j.jid.2021.10.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 10/13/2021] [Accepted: 10/20/2021] [Indexed: 11/22/2022]
|
22
|
Wang J, Zhang XY, Xu DY. Zebrafish xenograft model for studying the function of lncRNA SNHG4 in the proliferation and migration of colorectal cancer. J Gastrointest Oncol 2022; 13:210-220. [PMID: 35284103 PMCID: PMC8899727 DOI: 10.21037/jgo-21-832] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/30/2022] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND The zebrafish xenograft model has become a reliable in vivo model for human cancer research. Compared to a mouse model, the zebrafish xenograft has many advantages, including optical transparency, intuitive in vivo observation, and speed. Long noncoding RNAs (lncRNAs) have been identified as crucial regulatory factors in the progression of colorectal cancer (CRC). The biological function of lncRNA small nucleolar RNA host gene 4 (SNHG4) in CRC is still unclear. METHODS We analyzed the expression of SNHG4 in CRC patient samples by the Gene Expression Profiling Interactive Analysis (GEPIA) software. The quantitative real time-polymerase chain reaction (qRT-PCR) was used to verify in CRC cell lines. The colony formation assay was used to study the cell proliferation, and we used the transwell assay to detect the migration ability. Then the zebrafish xenograft models were used to confirm these roles of SNHG4 in vivo. Moreover, we detected epithelial mesenchymal transition (EMT) related genes by qRT-PCR. RESULTS We found the expression of SNHG4 was upregulated in CRC patient samples by analyzing GEPIA software, which was also verified in CRC cell lines. We also found that silencing SNHG4 inhibited the proliferation and migration of CRC cells, and its roles were verified in zebrafish xenografts in vivo. Further, we found that the expression of E-cadherin was significantly upregulated and N-cadherin was downregulated when knocking-down SNHG4 in CRC cells. CONCLUSIONS Our findings demonstrated that SNHG4 played oncogenic roles in CRC, which could be a potential target for treatment of CRC patients, and the results strongly revealed that zebrafish xenograft could be used for functional research of lncRNAs in human cancer.
Collapse
Affiliation(s)
- Jian Wang
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
- Department of General Surgery, The Affiliated Huai’an Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiao-Yu Zhang
- Department of General Surgery, The Affiliated Huai’an Hospital of Xuzhou Medical University, Xuzhou, China
| | - Dong-Yan Xu
- Department of Gastroenterology, The Affiliated Huai’an Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
23
|
Somasagara RR, Leung T. Zebrafish Xenograft Model to Study Human Cancer. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2413:45-53. [PMID: 35044653 DOI: 10.1007/978-1-0716-1896-7_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The zebrafish, Danio rerio, has been an important animal model for cancer research over the last decade. The capability of a high-throughput screen in zebrafish and a wide range of pharmacologically active compounds elicit physiological responses in zebrafish embryos comparable to those in mammalian systems, making zebrafish ideal for identifying clinically relevant drug targets and compounds that regulate tumor progression. The zebrafish model is suitable for patient-derived xenograft (pdx) and large-scale screening of lead compounds against specific malignancies. This established vertebrate model has many advantages, including fast response time, cost efficiency for drug testing, efficient manipulation of the host microenvironment by genetic tools, suitable for small molecule drug screening in high-throughput setting, easy maintenance, transparency for easy observation, high fecundity, and rapid generation time. The zebrafish model is a good alternative in vivo model to mammals for robust testing of drug candidates for cancer therapy.
Collapse
Affiliation(s)
- Ranganatha R Somasagara
- The Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, North Carolina Research Campus, Kannapolis, NC, USA
| | - TinChung Leung
- The Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, North Carolina Research Campus, Kannapolis, NC, USA. .,Department of Biological and Biomedical Sciences, North Carolina Central University, Durham, NC, USA.
| |
Collapse
|
24
|
Weiss JM, Lumaquin-Yin D, Montal E, Suresh S, Leonhardt CS, White RM. Shifting the focus of zebrafish toward a model of the tumor microenvironment. eLife 2022; 11:69703. [PMID: 36538362 PMCID: PMC9767465 DOI: 10.7554/elife.69703] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 11/21/2022] [Indexed: 12/29/2022] Open
Abstract
Cancer cells exist in a complex ecosystem with numerous other cell types in the tumor microenvironment (TME). The composition of this tumor/TME ecosystem will vary at each anatomic site and affects phenotypes such as initiation, metastasis, and drug resistance. A mechanistic understanding of the large number of cell-cell interactions between tumor and TME requires models that allow us to both characterize as well as genetically perturb this complexity. Zebrafish are a model system optimized for this problem, because of the large number of existing cell-type-specific drivers that can label nearly any cell in the TME. These include stromal cells, immune cells, and tissue resident normal cells. These cell-type-specific promoters/enhancers can be used to drive fluorophores to facilitate imaging and also CRISPR cassettes to facilitate perturbations. A major advantage of the zebrafish is the ease by which large numbers of TME cell types can be studied at once, within the same animal. While these features make the zebrafish well suited to investigate the TME, the model has important limitations, which we also discuss. In this review, we describe the existing toolset for studying the TME using zebrafish models of cancer and highlight unique biological insights that can be gained by leveraging this powerful resource.
Collapse
Affiliation(s)
- Joshua M Weiss
- Weill-Cornel Medical College, Tri-Institutional M.D./Ph.D. ProgramNew YorkUnited States
| | - Dianne Lumaquin-Yin
- Weill-Cornel Medical College, Tri-Institutional M.D./Ph.D. ProgramNew YorkUnited States
| | - Emily Montal
- Memorial Sloan Kettering Cancer Center, Department of Cancer Biology & GeneticsNew YorkUnited States
| | - Shruthy Suresh
- Memorial Sloan Kettering Cancer Center, Department of Cancer Biology & GeneticsNew YorkUnited States
| | - Carl S Leonhardt
- Memorial Sloan Kettering Cancer Center, Department of Cancer Biology & GeneticsNew YorkUnited States
| | - Richard M White
- Memorial Sloan Kettering Cancer Center, Department of Cancer Biology & GeneticsNew YorkUnited States,Department of Medicine, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| |
Collapse
|
25
|
Mendonça-Gomes JM, Valverde TM, Martins TMDM, Charlie-Silva I, Padovani BN, Fénero CM, da Silva EM, Domingues RZ, Melo-Hoyos DC, Corrêa-Junior JD, Câmara NOS, Góes AM, Gomes DA. Long-term dexamethasone treatment increases the engraftment efficiency of human breast cancer cells in adult zebrafish. FISH AND SHELLFISH IMMUNOLOGY REPORTS 2021; 2:100007. [DOI: 10.1016/j.fsirep.2021.100007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/03/2021] [Accepted: 03/03/2021] [Indexed: 01/03/2023] Open
|
26
|
Saggioro M, D'Agostino S, Gallo A, Crotti S, D'Aronco S, Corallo D, Veltri G, Martinez G, Grigoletto A, Tolomeo AM, Tafuro G, Agostini M, Aveic S, Serafin V, Semenzato A, Pasut G, Pozzobon M. A rhabdomyosarcoma hydrogel model to unveil cell-extracellular matrix interactions. Biomater Sci 2021; 10:124-137. [PMID: 34796888 DOI: 10.1039/d1bm00929j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Three-dimensional (3D) culture systems have progressively attracted attention given their potential to overcome limitations of classical 2D in vitro systems. Among different supports for 3D cell culture, hydrogels (HGs) offer important advantages such as tunable mechanical and biological properties. Here, a biocompatible hyaluronic acid-polyethylene glycol HG was developed to explore the pro-migratory behavior of alveolar rhabdomyosarcoma (ARMS) cells. Proteomic analysis of ARMS xenografts unveiled the composition of the extracellular matrix (ECM) elucidating the most representative proteins. In parallel, HGs were obtained by the combination of a thiol-containing hyaluronic acid derivative and different polyethylene glycol (PEG) dimaleimide polymers. The selection of the optimal HG for ARMS cell growth was made based on degradation time, swelling, and cell distribution. Rheology measures and mechanical properties were assessed in the presence or absence of ECM proteins (collagen type I and fibronectin), as well as viability tests and cell distribution analysis. The role of ITGA5, the receptor of fibronectin, in determining ARMS cell migration was validated in vitro upon ITGA5 silencing. In vivo, cell dissemination and the capacity for engrafting were validated after injecting ARMS cell populations enriched for the level of ITGA5 in zebrafish embryos. To study the interactions with ARMS-specific ECM proteins (HG + P), the key players from the Rho and heat-shock pathways were investigated by reverse phase protein array (RPPA). Our data suggest that the developed 3D ARMS model is useful for identifying potential physical hallmarks that allow cancer cells to resist therapy, escape from the immune-system and increase dissemination.
Collapse
Affiliation(s)
- Mattia Saggioro
- Stem Cells and Regenerative Medicine Lab, Institute of Pediatric Research Città della Speranza, 35129 Padova, Italy. .,Department of Women and Children Health, University of Padova, 35127 Padova, Italy
| | - Stefania D'Agostino
- Stem Cells and Regenerative Medicine Lab, Institute of Pediatric Research Città della Speranza, 35129 Padova, Italy. .,Department of Women and Children Health, University of Padova, 35127 Padova, Italy
| | - Anna Gallo
- Stem Cells and Regenerative Medicine Lab, Institute of Pediatric Research Città della Speranza, 35129 Padova, Italy. .,Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy.
| | - Sara Crotti
- NIB Lab Institute of Pediatric Research Città della Speranza, 35129 Padova, Italy
| | - Sara D'Aronco
- NIB Lab Institute of Pediatric Research Città della Speranza, 35129 Padova, Italy
| | - Diana Corallo
- Laboratory of Target Discovery and Biology of Neuroblastoma, Institute of Pediatric Research Città della Speranza, 35129 Padova, Italy
| | - Giulia Veltri
- Department of Women and Children Health, University of Padova, 35127 Padova, Italy.,Oncohematology Laboratory, Institute of Pediatric Research Città della Speranza, 35129 Padova, Italy
| | - Gabriele Martinez
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy.
| | - Antonella Grigoletto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy.
| | - Anna Maria Tolomeo
- Department of Women and Children Health, University of Padova, 35127 Padova, Italy.,L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), 35129 Padova, Italy
| | - Giovanni Tafuro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy.
| | - Marco Agostini
- First Surgical Clinic Section, Department of Surgical, Oncological and Gastroenterological Sciences, Padova University, 35128 Padova, Italy.,NIB Lab Institute of Pediatric Research Città della Speranza, 35129 Padova, Italy.,L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), 35129 Padova, Italy
| | - Sanja Aveic
- Laboratory of Target Discovery and Biology of Neuroblastoma, Institute of Pediatric Research Città della Speranza, 35129 Padova, Italy.,Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Aachen, Germany
| | - Valentina Serafin
- Department of Women and Children Health, University of Padova, 35127 Padova, Italy.,Oncohematology Laboratory, Institute of Pediatric Research Città della Speranza, 35129 Padova, Italy
| | - Alessandra Semenzato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy.
| | - Gianfranco Pasut
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy.
| | - Michela Pozzobon
- Stem Cells and Regenerative Medicine Lab, Institute of Pediatric Research Città della Speranza, 35129 Padova, Italy. .,Department of Women and Children Health, University of Padova, 35127 Padova, Italy
| |
Collapse
|
27
|
Molina B, Chavez J, Grainger S. Zebrafish models of acute leukemias: Current models and future directions. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2021; 10:e400. [PMID: 33340278 PMCID: PMC8213871 DOI: 10.1002/wdev.400] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/02/2020] [Accepted: 11/09/2020] [Indexed: 12/19/2022]
Abstract
Acute myeloid leukemias (AML) and acute lymphoid leukemias (ALL) are heterogenous diseases encompassing a wide array of genetic mutations with both loss and gain of function phenotypes. Ultimately, these both result in the clonal overgrowth of blast cells in the bone marrow, peripheral blood, and other tissues. As a consequence of this, normal hematopoietic stem cell function is severely hampered. Technologies allowing for the early detection of genetic alterations and understanding of these varied molecular pathologies have helped to advance our treatment regimens toward personalized targeted therapies. In spite of this, both AML and ALL continue to be a major cause of morbidity and mortality worldwide, in part because molecular therapies for the plethora of genetic abnormalities have not been developed. This underscores the current need for better model systems for therapy development. This article reviews the current zebrafish models of AML and ALL and discusses how novel gene editing tools can be implemented to generate better models of acute leukemias. This article is categorized under: Adult Stem Cells, Tissue Renewal, and Regeneration > Stem Cells and Disease Technologies > Perturbing Genes and Generating Modified Animals.
Collapse
Affiliation(s)
- Brandon Molina
- Biology Department, San Diego State University, San Diego, California, USA
| | - Jasmine Chavez
- Biology Department, San Diego State University, San Diego, California, USA
| | - Stephanie Grainger
- Biology Department, San Diego State University, San Diego, California, USA
| |
Collapse
|
28
|
Lou Y, Jiang Y, Liang Z, Liu B, Li T, Zhang D. Role of RhoC in cancer cell migration. Cancer Cell Int 2021; 21:527. [PMID: 34627249 PMCID: PMC8502390 DOI: 10.1186/s12935-021-02234-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/27/2021] [Indexed: 12/20/2022] Open
Abstract
Migration is one of the five major behaviors of cells. Although RhoC-a classic member of the Rho gene family-was first identified in 1985, functional RhoC data have only been widely reported in recent years. Cell migration involves highly complex signaling mechanisms, in which RhoC plays an essential role. Cell migration regulated by RhoC-of which the most well-known function is its role in cancer metastasis-has been widely reported in breast, gastric, colon, bladder, prostate, lung, pancreatic, liver, and other cancers. Our review describes the role of RhoC in various types of cell migration. The classic two-dimensional cell migration cycle constitutes cell polarization, adhesion regulation, cell contraction and tail retraction, most of which are modulated by RhoC. In the three-dimensional cell migration model, amoeboid migration is the most classic and well-studied model. Here, RhoC modulates the formation of membrane vesicles by regulating myosin II, thereby affecting the rate and persistence of amoeba-like migration. To the best of our knowledge, this review is the first to describe the role of RhoC in all cell migration processes. We believe that understanding the detail of RhoC-regulated migration processes will help us better comprehend the mechanism of cancer metastasis. This will contribute to the study of anti-metastatic treatment approaches, aiding in the identification of new intervention targets for therapeutic or genetic transformational purposes.
Collapse
Affiliation(s)
- Yingyue Lou
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yuhan Jiang
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhen Liang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Bingzhang Liu
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Tian Li
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, Jilin, China.
| | - Duo Zhang
- Department of Plastic and Reconstructive Surgery, The First Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
29
|
Crosas-Molist E, Samain R, Kohlhammer L, Orgaz J, George S, Maiques O, Barcelo J, Sanz-Moreno V. RhoGTPase Signalling in Cancer Progression and Dissemination. Physiol Rev 2021; 102:455-510. [PMID: 34541899 DOI: 10.1152/physrev.00045.2020] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Rho GTPases are a family of small G proteins that regulate a wide array of cellular processes related to their key roles controlling the cytoskeleton. On the other hand, cancer is a multi-step disease caused by the accumulation of genetic mutations and epigenetic alterations, from the initial stages of cancer development when cells in normal tissues undergo transformation, to the acquisition of invasive and metastatic traits, responsible for a large number of cancer related deaths. In this review, we discuss the role of Rho GTPase signalling in cancer in every step of disease progression. Rho GTPases contribute to tumour initiation and progression, by regulating proliferation and apoptosis, but also metabolism, senescence and cell stemness. Rho GTPases play a major role in cell migration, and in the metastatic process. They are also involved in interactions with the tumour microenvironment and regulate inflammation, contributing to cancer progression. After years of intensive research, we highlight the importance of relevant models in the Rho GTPase field, and we reflect on the therapeutic opportunities arising for cancer patients.
Collapse
Affiliation(s)
- Eva Crosas-Molist
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Remi Samain
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Leonie Kohlhammer
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Jose Orgaz
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.,Instituto de Investigaciones Biomédicas 'Alberto Sols', CSIC-UAM, 28029, Madrid, Spain
| | - Samantha George
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Oscar Maiques
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Jaume Barcelo
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | | |
Collapse
|
30
|
Umans RA, Ten Kate M, Pollock C, Sontheimer H. Fishing for Contact: Modeling Perivascular Glioma Invasion in the Zebrafish Brain. ACS Pharmacol Transl Sci 2021; 4:1295-1305. [PMID: 34423267 DOI: 10.1021/acsptsci.0c00129] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Indexed: 12/16/2022]
Abstract
Glioblastoma multiforme (GBM) is a highly invasive, central nervous system (CNS) cancer for which there is no cure. Invading tumor cells evade treatment, limiting the efficacy of the current standard of care regimen. Understanding the underlying invasive behaviors that support tumor growth may allow for generation of novel GBM therapies. Zebrafish (Danio rerio) are attractive for genetics and live imaging and have, in recent years, emerged as a model system suitable for cancer biology research. While other groups have studied CNS tumors using zebrafish, few have concentrated on the invasive behaviors supporting the development of these diseases. Previous studies demonstrated that one of the main mechanisms of GBM invasion is perivascular invasion, i.e., single tumor cell migration along blood vessels. Here, we characterize phenotypes, methodology, and potential therapeutic avenues for utilizing zebrafish to model perivascular GBM invasion. Using patient-derived xenolines or an adherent cell line, we demonstrate tumor expansion within the zebrafish brain. Within 24-h postintracranial injection, D54-MG-tdTomato glioma cells produce fingerlike projections along the zebrafish brain vasculature. As few as 25 GBM cells were sufficient to promote single cell vessel co-option. Of note, these tumor-vessel interactions are CNS specific and do not occur on pre-existing blood vessels when injected into the animal's peripheral tissue. Tumor-vessel interactions increase over time and can be pharmacologically disrupted through inhibition of Wnt signaling. Therefore, zebrafish serve as a favorable model system to study perivascular glioma invasion, one of the deadly characteristics that make GBM so difficult to treat.
Collapse
Affiliation(s)
- Robyn A Umans
- Center for Glial Biology in Health, Disease, and Cancer, The Fralin Biomedical Research Institute at VTC, Roanoke, Virginia 24016, United States
| | - Mattie Ten Kate
- School of Neuroscience, Virginia Tech, Sandy Hall, 210 Drillfield Drive, Blacksburg, Virginia 24061, United States
| | - Carolyn Pollock
- School of Neuroscience, Virginia Tech, Sandy Hall, 210 Drillfield Drive, Blacksburg, Virginia 24061, United States
| | - Harald Sontheimer
- Center for Glial Biology in Health, Disease, and Cancer, The Fralin Biomedical Research Institute at VTC, Roanoke, Virginia 24016, United States.,School of Neuroscience, Virginia Tech, Sandy Hall, 210 Drillfield Drive, Blacksburg, Virginia 24061, United States
| |
Collapse
|
31
|
Anvari S, Osei E, Maftoon N. Interactions of platelets with circulating tumor cells contribute to cancer metastasis. Sci Rep 2021; 11:15477. [PMID: 34326373 PMCID: PMC8322323 DOI: 10.1038/s41598-021-94735-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/09/2021] [Indexed: 02/07/2023] Open
Abstract
Recent studies have suggested that platelets have a crucial role in enhancing the survival of circulating tumor cells in the bloodstream and aggravating cancer metastasis. The main function of platelets is to bind to the sites of the damaged vessels to stop bleeding. However, in cancer patients, activated platelets adhere to circulating tumor cells and exacerbate metastatic spreading. Several hypotheses have been proposed about the platelet-cancer cell interactions, but the underlying mechanisms of these interactions are not completely understood yet. In this work, we quantitatively investigated the interactions between circulating tumor cells, red blood cells, platelets, plasma flow and microvessel walls via computational modelling at the cellular scale. Our highly detailed computational model allowed us to understand and quantitatively explain the role of platelets in deformation, adhesion and survival of tumor cells in their active arrest to the endothelium.
Collapse
Affiliation(s)
- Sina Anvari
- Department of Systems Design Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
- Centre for Bioengineering and Biotechnology, University of Waterloo, Waterloo, ON, Canada
| | - Ernest Osei
- Department of Systems Design Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
- Department of Medical Physics, Grand River Regional Cancer Centre, Kitchener, ON, Canada
- Department of Physics and Astronomy, University of Waterloo, Waterloo, ON, Canada
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Nima Maftoon
- Department of Systems Design Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada.
- Centre for Bioengineering and Biotechnology, University of Waterloo, Waterloo, ON, Canada.
| |
Collapse
|
32
|
Pensado-López A, Fernández-Rey J, Reimunde P, Crecente-Campo J, Sánchez L, Torres Andón F. Zebrafish Models for the Safety and Therapeutic Testing of Nanoparticles with a Focus on Macrophages. NANOMATERIALS 2021; 11:nano11071784. [PMID: 34361170 PMCID: PMC8308170 DOI: 10.3390/nano11071784] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 12/11/2022]
Abstract
New nanoparticles and biomaterials are increasingly being used in biomedical research for drug delivery, diagnostic applications, or vaccines, and they are also present in numerous commercial products, in the environment and workplaces. Thus, the evaluation of the safety and possible therapeutic application of these nanomaterials has become of foremost importance for the proper progress of nanotechnology. Due to economical and ethical issues, in vitro and in vivo methods are encouraged for the testing of new compounds and/or nanoparticles, however in vivo models are still needed. In this scenario, zebrafish (Danio rerio) has demonstrated potential for toxicological and pharmacological screenings. Zebrafish presents an innate immune system, from early developmental stages, with conserved macrophage phenotypes and functions with respect to humans. This fact, combined with the transparency of zebrafish, the availability of models with fluorescently labelled macrophages, as well as a broad variety of disease models offers great possibilities for the testing of new nanoparticles. Thus, with a particular focus on macrophage-nanoparticle interaction in vivo, here, we review the studies using zebrafish for toxicological and biodistribution testing of nanoparticles, and also the possibilities for their preclinical evaluation in various diseases, including cancer and autoimmune, neuroinflammatory, and infectious diseases.
Collapse
Affiliation(s)
- Alba Pensado-López
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (J.F.-R.)
- Center for Research in Molecular Medicine & Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain;
| | - Juan Fernández-Rey
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (J.F.-R.)
- Center for Research in Molecular Medicine & Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain;
| | - Pedro Reimunde
- Department of Physiotherapy, Medicine and Biomedical Sciences, Universidade da Coruña, Campus de Oza, 15006 A Coruña, Spain;
- Department of Neurosurgery, Hospital Universitario Lucus Augusti, 27003 Lugo, Spain
| | - José Crecente-Campo
- Center for Research in Molecular Medicine & Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain;
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (J.F.-R.)
- Correspondence: (L.S.); (F.T.A.)
| | - Fernando Torres Andón
- Center for Research in Molecular Medicine & Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain;
- Correspondence: (L.S.); (F.T.A.)
| |
Collapse
|
33
|
Yankaskas CL, Bera K, Stoletov K, Serra SA, Carrillo-Garcia J, Tuntithavornwat S, Mistriotis P, Lewis JD, Valverde MA, Konstantopoulos K. The fluid shear stress sensor TRPM7 regulates tumor cell intravasation. SCIENCE ADVANCES 2021; 7:7/28/eabh3457. [PMID: 34244134 PMCID: PMC8270498 DOI: 10.1126/sciadv.abh3457] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/28/2021] [Indexed: 05/09/2023]
Abstract
Tumor cell intravasation preferentially occurs in regions of low fluid shear because high shear is detrimental to tumor cells. Here, we describe a molecular mechanism by which cells avoid high shear during intravasation. The transition from migration to intravasation was modeled using a microfluidic device where cells migrating inside longitudinal tissue-like microchannels encounter an orthogonal channel in which fluid flow induces physiological shear stresses. This approach was complemented with intravital microscopy, patch-clamp, and signal transduction imaging techniques. Fluid shear-induced activation of the transient receptor potential melastatin 7 (TRPM7) channel promotes extracellular calcium influx, which then activates RhoA/myosin-II and calmodulin/IQGAP1/Cdc42 pathways to coordinate reversal of migration direction, thereby avoiding shear stress. Cells displaying higher shear sensitivity due to higher TRPM7 activity levels intravasate less efficiently and establish less invasive metastatic lesions. This study provides a mechanistic interpretation for the role of shear stress and its sensor, TRPM7, in tumor cell intravasation.
Collapse
Affiliation(s)
- Christopher L Yankaskas
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Kaustav Bera
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | | | - Selma A Serra
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Julia Carrillo-Garcia
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Soontorn Tuntithavornwat
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Panagiotis Mistriotis
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA
| | - John D Lewis
- Department of Oncology, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Miguel A Valverde
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA.
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
34
|
Vasilaki D, Bakopoulou A, Tsouknidas A, Johnstone E, Michalakis K. Biophysical interactions between components of the tumor microenvironment promote metastasis. Biophys Rev 2021; 13:339-357. [PMID: 34168685 PMCID: PMC8214652 DOI: 10.1007/s12551-021-00811-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 05/03/2021] [Indexed: 02/07/2023] Open
Abstract
During metastasis, tumor cells need to adapt to their dynamic microenvironment and modify their mechanical properties in response to both chemical and mechanical stimulation. Physical interactions occur between cancer cells and the surrounding matrix including cell movements and cell shape alterations through the process of mechanotransduction. The latter describes the translation of external mechanical cues into intracellular biochemical signaling. Reorganization of both the cytoskeleton and the extracellular matrix (ECM) plays a critical role in these spreading steps. Migrating tumor cells show increased motility in order to cross the tumor microenvironment, migrate through ECM and reach the bloodstream to the metastatic site. There are specific factors affecting these processes, as well as the survival of circulating tumor cells (CTC) in the blood flow until they finally invade the secondary tissue to form metastasis. This review aims to study the mechanisms of metastasis from a biomechanical perspective and investigate cell migration, with a focus on the alterations in the cytoskeleton through this journey and the effect of biologic fluids on metastasis. Understanding of the biophysical mechanisms that promote tumor metastasis may contribute successful therapeutic approaches in the fight against cancer.
Collapse
Affiliation(s)
- Dimitra Vasilaki
- Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Athina Bakopoulou
- Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Alexandros Tsouknidas
- Laboratory for Biomaterials and Computational Mechanics, Department of Mechanical Engineering, University of Western Macedonia, Kozani, Greece
| | | | - Konstantinos Michalakis
- Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
- Division of Graduate Prosthodontics, Tufts University School of Dental Medicine, Boston, MA USA
- University of Oxford, Oxford, UK
| |
Collapse
|
35
|
Saraiva SM, Gutiérrez-Lovera C, Martínez-Val J, Lores S, Bouzo BL, Díez-Villares S, Alijas S, Pensado-López A, Vázquez-Ríos AJ, Sánchez L, de la Fuente M. Edelfosine nanoemulsions inhibit tumor growth of triple negative breast cancer in zebrafish xenograft model. Sci Rep 2021; 11:9873. [PMID: 33972572 PMCID: PMC8110995 DOI: 10.1038/s41598-021-87968-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/29/2021] [Indexed: 12/18/2022] Open
Abstract
Triple negative breast cancer (TNBC) is known for being very aggressive, heterogeneous and highly metastatic. The standard of care treatment is still chemotherapy, with adjacent toxicity and low efficacy, highlighting the need for alternative and more effective therapeutic strategies. Edelfosine, an alkyl-lysophospholipid, has proved to be a promising therapy for several cancer types, upon delivery in lipid nanoparticles. Therefore, the objective of this work was to explore the potential of edelfosine for the treatment of TNBC. Edelfosine nanoemulsions (ET-NEs) composed by edelfosine, Miglyol 812 and phosphatidylcholine as excipients, due to their good safety profile, presented an average size of about 120 nm and a neutral zeta potential, and were stable in biorelevant media. The ability of ET-NEs to interrupt tumor growth in TNBC was demonstrated both in vitro, using a highly aggressive and invasive TNBC cell line, and in vivo, using zebrafish embryos. Importantly, ET-NEs were able to penetrate through the skin barrier of MDA-MB 231 xenografted zebrafish embryos, into the yolk sac, leading to an effective decrease of highly aggressive and invasive tumoral cells' proliferation. Altogether the results demonstrate the potential of ET-NEs for the development of new therapeutic approaches for TNBC.
Collapse
Affiliation(s)
- Sofia M Saraiva
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Spain
- Cancer Network Research (CIBERONC), Madrid, Spain
| | - Carlha Gutiérrez-Lovera
- Department of Zoology, Genetics and Physical Anthropology, Campus of Lugo, University of Santiago de Compostela, Lugo, Spain
| | - Jeannette Martínez-Val
- Department of Zoology, Genetics and Physical Anthropology, Campus of Lugo, University of Santiago de Compostela, Lugo, Spain
| | - Sainza Lores
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Spain
| | - Belén L Bouzo
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Spain
| | - Sandra Díez-Villares
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Spain
- Cancer Network Research (CIBERONC), Madrid, Spain
| | - Sandra Alijas
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Spain
| | - Alba Pensado-López
- Department of Zoology, Genetics and Physical Anthropology, Campus of Lugo, University of Santiago de Compostela, Lugo, Spain
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Abi Judit Vázquez-Ríos
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Spain
- Cancer Network Research (CIBERONC), Madrid, Spain
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Campus of Lugo, University of Santiago de Compostela, Lugo, Spain
| | - María de la Fuente
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Spain.
- Cancer Network Research (CIBERONC), Madrid, Spain.
| |
Collapse
|
36
|
Visualizing Extracellular Vesicles and Their Function in 3D Tumor Microenvironment Models. Int J Mol Sci 2021; 22:ijms22094784. [PMID: 33946403 PMCID: PMC8125158 DOI: 10.3390/ijms22094784] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) are cell-derived nanostructures that mediate intercellular communication by delivering complex signals in normal tissues and cancer. The cellular coordination required for tumor development and maintenance is mediated, in part, through EV transport of molecular cargo to resident and distant cells. Most studies on EV-mediated signaling have been performed in two-dimensional (2D) monolayer cell cultures, largely because of their simplicity and high-throughput screening capacity. Three-dimensional (3D) cell cultures can be used to study cell-to-cell and cell-to-matrix interactions, enabling the study of EV-mediated cellular communication. 3D cultures may best model the role of EVs in formation of the tumor microenvironment (TME) and cancer cell-stromal interactions that sustain tumor growth. In this review, we discuss EV biology in 3D culture correlates of the TME. This includes EV communication between cell types of the TME, differences in EV biogenesis and signaling associated with differing scaffold choices and in scaffold-free 3D cultures and cultivation of the premetastatic niche. An understanding of EV biogenesis and signaling within a 3D TME will improve culture correlates of oncogenesis, enable molecular control of the TME and aid development of drug delivery tools based on EV-mediated signaling.
Collapse
|
37
|
Gamble JT, Elson DJ, Greenwood JA, Tanguay RL, Kolluri SK. The Zebrafish Xenograft Models for Investigating Cancer and Cancer Therapeutics. BIOLOGY 2021; 10:biology10040252. [PMID: 33804830 PMCID: PMC8063817 DOI: 10.3390/biology10040252] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 03/17/2021] [Indexed: 02/06/2023]
Abstract
Simple Summary The identification and development of new anti-cancer drugs requires extensive testing in animal models to establish safety and efficacy of drug candidates. The transplantation of human tumor tissue into mouse (tumor xenografts) is commonly used to study cancer progression and to test potential drugs for their anti-cancer activity. Mouse models do not afford the ability to test a large number of drug candidates quickly as it takes several weeks to conduct these experiments. In contrast, tumor xenograft studies in zebrafish provide an efficient platform for rapid testing of safety and efficacy in less than two weeks. Abstract In order to develop new cancer therapeutics, rapid, reliable, and relevant biological models are required to screen and validate drug candidates for both efficacy and safety. In recent years, the zebrafish (Danio rerio) has emerged as an excellent model organism suited for these goals. Larval fish or immunocompromised adult fish are used to engraft human cancer cells and serve as a platform for screening potential drug candidates. With zebrafish sharing ~80% of disease-related orthologous genes with humans, they provide a low cost, high-throughput alternative to mouse xenografts that is relevant to human biology. In this review, we provide background on the methods and utility of zebrafish xenograft models in cancer research.
Collapse
Affiliation(s)
- John T. Gamble
- Department of Biochemistry & Biophysics, Oregon State University, Corvallis, OR 97331, USA;
| | - Daniel J. Elson
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA;
| | - Juliet A. Greenwood
- School of Mathematics and Natural Sciences, Arizona State University, Scotsdale, AZ 85257, USA;
| | - Robyn L. Tanguay
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA;
| | - Siva K. Kolluri
- Cancer Research Laboratory, Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA;
- Correspondence:
| |
Collapse
|
38
|
Sonay AY, Kalyviotis K, Yaganoglu S, Unsal A, Konantz M, Teulon C, Lieberwirth I, Sieber S, Jiang S, Behzadi S, Crespy D, Landfester K, Roke S, Lengerke C, Pantazis P. Biodegradable Harmonophores for Targeted High-Resolution In Vivo Tumor Imaging. ACS NANO 2021; 15:4144-4154. [PMID: 33630589 PMCID: PMC8023799 DOI: 10.1021/acsnano.0c10634] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 02/05/2021] [Indexed: 05/31/2023]
Abstract
Optical imaging probes have played a major role in detecting and monitoring a variety of diseases. In particular, nonlinear optical imaging probes, such as second harmonic generating (SHG) nanoprobes, hold great promise as clinical contrast agents, as they can be imaged with little background signal and unmatched long-term photostability. As their chemical composition often includes transition metals, the use of inorganic SHG nanoprobes can raise long-term health concerns. Ideally, contrast agents for biomedical applications should be degraded in vivo without any long-term toxicological consequences to the organism. Here, we developed biodegradable harmonophores (bioharmonophores) that consist of polymer-encapsulated, self-assembling peptides that generate a strong SHG signal. When functionalized with tumor cell surface markers, these reporters can target single cancer cells with high detection sensitivity in zebrafish embryos in vivo. Thus, bioharmonophores will enable an innovative approach to cancer treatment using targeted high-resolution optical imaging for diagnostics and therapy.
Collapse
Affiliation(s)
- Ali Yasin Sonay
- Department
of Biosystems Science and Engineering (D-BSSE), Eidgenössische Technische Hochschule (ETH) Zurich, 4058 Basel, Switzerland
| | - Konstantinos Kalyviotis
- Department
of Bioengineering, Imperial College London, South Kensington Campus, London SW7 2AZ, U.K.
| | - Sine Yaganoglu
- Department
of Biosystems Science and Engineering (D-BSSE), Eidgenössische Technische Hochschule (ETH) Zurich, 4058 Basel, Switzerland
| | - Aysen Unsal
- Department
of Bioengineering, Imperial College London, South Kensington Campus, London SW7 2AZ, U.K.
| | - Martina Konantz
- Department
of Biomedicine, University Hospital Basel
and University of Basel, 4031 Basel, Switzerland
| | - Claire Teulon
- Laboratory
for Fundamental BioPhotonics, Institute of Bioengineering, School
of Engineering, École Polytechnique
Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | | | - Sandro Sieber
- Division
of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, 4031 Basel, Switzerland
| | - Shuai Jiang
- Max
Planck Institute for Polymer Research, 55128 Mainz, Germany
| | - Shahed Behzadi
- Max
Planck Institute for Polymer Research, 55128 Mainz, Germany
| | - Daniel Crespy
- Max
Planck Institute for Polymer Research, 55128 Mainz, Germany
- Department
of Materials Science and Engineering, School of Molecular Science
and Engineering, Vidyasirimedhi Institute
of Science and Technology (VISTEC), Rayong 21210, Thailand
| | | | - Sylvie Roke
- Laboratory
for Fundamental BioPhotonics, Institute of Bioengineering, School
of Engineering, École Polytechnique
Fédérale de Lausanne, 1015 Lausanne, Switzerland
- Institute
of Materials Science and Engineering, School of Engineering, École Polytechnique Fédérale
de Lausanne, 1015 Lausanne, Switzerland
- Lausanne
Centre for Ultrafast Science, École
Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Claudia Lengerke
- Department
of Biomedicine, University Hospital Basel
and University of Basel, 4031 Basel, Switzerland
- Division
of Hematology, University Hospital Basel, 4031 Basel, Switzerland
| | - Periklis Pantazis
- Department
of Biosystems Science and Engineering (D-BSSE), Eidgenössische Technische Hochschule (ETH) Zurich, 4058 Basel, Switzerland
- Department
of Bioengineering, Imperial College London, South Kensington Campus, London SW7 2AZ, U.K.
| |
Collapse
|
39
|
Her ZP, Yeo KS, Howe C, Levee T, Zhu S. Zebrafish Model of Neuroblastoma Metastasis. J Vis Exp 2021. [PMID: 33779609 DOI: 10.3791/62416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Zebrafish has emerged as an important animal model to study human diseases, especially cancer. Along with the robust transgenic and genome editing technologies applied in zebrafish modeling, the ease of maintenance, high-yield productivity, and powerful live imaging altogether make the zebrafish a valuable model system to study metastasis and cellular and molecular bases underlying this process in vivo. The first zebrafish neuroblastoma (NB) model of metastasis was developed by overexpressing two oncogenes, MYCN and LMO1, under control of the dopamine-beta-hydroxylase (dβh) promoter. Co-overexpressed MYCN and LMO1 led to the reduced latency and increased penetrance of neuroblastomagenesis, as well as accelerated distant metastasis of tumor cells. This new model reliably reiterates many key features of human metastatic NB, including involvement of clinically relevant and metastasis-associated genetic alterations; natural and spontaneous development of metastasis in vivo; and conserved sites of metastases. Therefore, the zebrafish model possesses unique advantages to dissect the complex process of tumor metastasis in vivo.
Collapse
Affiliation(s)
- Zuag Paj Her
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic Cancer Center
| | - Kok Siong Yeo
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic Cancer Center
| | - Cassie Howe
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic Cancer Center
| | - Taylor Levee
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic Cancer Center
| | - Shizhen Zhu
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Mayo Clinic Cancer Center;
| |
Collapse
|
40
|
Somasagara RR, Huang X, Xu C, Haider J, Serody JS, Armistead PM, Leung T. Targeted therapy of human leukemia xenografts in immunodeficient zebrafish. Sci Rep 2021; 11:5715. [PMID: 33707624 PMCID: PMC7952715 DOI: 10.1038/s41598-021-85141-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 02/25/2021] [Indexed: 01/05/2023] Open
Abstract
Personalized medicine holds tremendous promise for improving safety and efficacy of drug therapies by optimizing treatment regimens. Rapidly developed patient-derived xenografts (pdx) could be a helpful tool for analyzing the effect of drugs against an individual's tumor by growing the tumor in an immunodeficient animal. Severe combined immunodeficiency (SCID) mice enable efficient in vivo expansion of vital tumor cells and generation of personalized xenografts. However, they are not amenable to large-scale rapid screening, which is critical in identifying new compounds from large compound libraries. The development of a zebrafish model suitable for pdx could facilitate large-scale screening of drugs targeted against specific malignancies. Here, we describe a novel strategy for establishing a zebrafish model for drug testing in leukemia xenografts. We used chronic myelogenous leukemia and acute myeloid leukemia for xenotransplantation into SCID zebrafish to evaluate drug screening protocols. We showed the in vivo efficacy of the ABL inhibitor imatinib, MEK inhibitor U0126, cytarabine, azacitidine and arsenic trioxide. We performed corresponding in vitro studies, demonstrating that combination of MEK- and FLT3-inhibitors exhibit an enhanced effect in vitro. We further evaluated the feasibility of zebrafish for transplantation of primary human hematopoietic cells that can survive at 15 day-post-fertilization. Our results provide critical insights to guide development of high-throughput platforms for evaluating leukemia.
Collapse
Affiliation(s)
- Ranganatha R Somasagara
- The Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, North Carolina Research Campus, Kannapolis, NC, 28081, USA
| | - Xiaoyan Huang
- The Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, North Carolina Research Campus, Kannapolis, NC, 28081, USA
| | - Chunyu Xu
- The Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, North Carolina Research Campus, Kannapolis, NC, 28081, USA
| | - Jamil Haider
- The Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, North Carolina Research Campus, Kannapolis, NC, 28081, USA
| | - Jonathan S Serody
- Division of Hematology/Oncology, Department of Medicine, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Paul M Armistead
- Division of Hematology/Oncology, Department of Medicine, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - TinChung Leung
- The Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, North Carolina Research Campus, Kannapolis, NC, 28081, USA. .,Department of Biological & Biomedical Sciences, North Carolina Central University, Durham, NC, 27707, USA.
| |
Collapse
|
41
|
Cheng X, Cheng K. Visualizing cancer extravasation: from mechanistic studies to drug development. Cancer Metastasis Rev 2021; 40:71-88. [PMID: 33156478 PMCID: PMC7897269 DOI: 10.1007/s10555-020-09942-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023]
Abstract
Metastasis is a multistep process that accounts for the majority of cancer-related death. By the end of metastasize dissemination, circulating tumor cells (CTC) need to extravasate the blood vessels at metastatic sites to form new colonization. Although cancer cell extravasation is a crucial step in cancer metastasis, it has not been successfully targeted by current anti-metastasis strategies due to the lack of a thorough understanding of the molecular mechanisms that regulate this process. This review focuses on recent progress in cancer extravasation visualization techniques, including the development of both in vitro and in vivo cancer extravasation models, that shed light on the underlying mechanisms. Specifically, multiple cancer extravasation stages, such as the adhesion to the endothelium and transendothelial migration, are successfully probed using these technologies. Moreover, the roles of different cell adhesive molecules, chemokines, and growth factors, as well as the mechanical factors in these stages are well illustrated. Deeper understandings of cancer extravasation mechanisms offer us new opportunities to escalate the discovery of anti-extravasation drugs and therapies and improve the prognosis of cancer patients.
Collapse
Affiliation(s)
- Xiao Cheng
- Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina at Chapel Hill, Raleigh, NC, USA
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, 27607, USA
| | - Ke Cheng
- Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina at Chapel Hill, Raleigh, NC, USA.
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, 27607, USA.
| |
Collapse
|
42
|
Póvoa V, Rebelo de Almeida C, Maia-Gil M, Sobral D, Domingues M, Martinez-Lopez M, de Almeida Fuzeta M, Silva C, Grosso AR, Fior R. Innate immune evasion revealed in a colorectal zebrafish xenograft model. Nat Commun 2021; 12:1156. [PMID: 33608544 PMCID: PMC7895829 DOI: 10.1038/s41467-021-21421-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 01/26/2021] [Indexed: 01/31/2023] Open
Abstract
Cancer immunoediting is a dynamic process of crosstalk between tumor cells and the immune system. Herein, we explore the fast zebrafish xenograft model to investigate the innate immune contribution to this process. Using multiple breast and colorectal cancer cell lines and zAvatars, we find that some are cleared (regressors) while others engraft (progressors) in zebrafish xenografts. We focus on two human colorectal cancer cells derived from the same patient that show contrasting engraftment/clearance profiles. Using polyclonal xenografts to mimic intra-tumor heterogeneity, we demonstrate that SW620_progressors can block clearance of SW480_regressors. SW480_regressors recruit macrophages and neutrophils more efficiently than SW620_progressors; SW620_progressors however, modulate macrophages towards a pro-tumoral phenotype. Genetic and chemical suppression of myeloid cells indicates that macrophages and neutrophils play a crucial role in clearance. Single-cell-transcriptome analysis shows a fast subclonal selection, with clearance of regressor subclones associated with IFN/Notch signaling and escaper-expanded subclones with enrichment of IL10 pathway. Overall, our work opens the possibility of using zebrafish xenografts as living biomarkers of the tumor microenvironment.
Collapse
Affiliation(s)
- Vanda Póvoa
- Champalimaud Centre for the Unknown, Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| | - Cátia Rebelo de Almeida
- Champalimaud Centre for the Unknown, Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| | - Mariana Maia-Gil
- Champalimaud Centre for the Unknown, Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| | - Daniel Sobral
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Micaela Domingues
- Champalimaud Centre for the Unknown, Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| | - Mayra Martinez-Lopez
- Champalimaud Centre for the Unknown, Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| | - Miguel de Almeida Fuzeta
- Champalimaud Centre for the Unknown, Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| | - Carlos Silva
- Champalimaud Centre for the Unknown, Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| | - Ana Rita Grosso
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Rita Fior
- Champalimaud Centre for the Unknown, Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal.
| |
Collapse
|
43
|
Chen X, Li Y, Yao T, Jia R. Benefits of Zebrafish Xenograft Models in Cancer Research. Front Cell Dev Biol 2021; 9:616551. [PMID: 33644052 PMCID: PMC7905065 DOI: 10.3389/fcell.2021.616551] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/11/2021] [Indexed: 12/14/2022] Open
Abstract
As a promising in vivo tool for cancer research, zebrafish have been widely applied in various tumor studies. The zebrafish xenograft model is a low-cost, high-throughput tool for cancer research that can be established quickly and requires only a small sample size, which makes it favorite among researchers. Zebrafish patient-derived xenograft (zPDX) models provide promising evidence for short-term clinical treatment. In this review, we discuss the characteristics and advantages of zebrafish, such as their transparent and translucent features, the use of vascular fluorescence imaging, the establishment of metastatic and intracranial orthotopic models, individual pharmacokinetics measurements, and tumor microenvironment. Furthermore, we introduce how these characteristics and advantages are applied other in tumor studies. Finally, we discuss the future direction of the use of zebrafish in tumor studies and provide new ideas for the application of it.
Collapse
Affiliation(s)
- Xingyu Chen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yongyun Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Tengteng Yao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| |
Collapse
|
44
|
Amawi H, Aljabali AAA, Boddu SHS, Amawi S, Obeid MA, Ashby CR, Tiwari AK. The use of zebrafish model in prostate cancer therapeutic development and discovery. Cancer Chemother Pharmacol 2021; 87:311-325. [PMID: 33392639 DOI: 10.1007/s00280-020-04211-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 11/26/2020] [Indexed: 12/24/2022]
Abstract
Zebrafish is now among the leading in vivo model for cancer research, including prostate cancer. They are an alternative economic model being used to study cancer development, proliferation, and metastasis. They can also be effectively utilized for the development of cancer drugs at all levels, including target validation, and high-throughput screening for possible lead molecules. In this review, we provide a comprehensive overview of the role of zebrafish as an in vivo model in prostate cancer research. Globally, prostate cancer is a leading cause of death in men. Although many molecular mechanisms have been identified as playing a role in the pathogenesis of prostate cancer, there is still a significant need to understand the initial events of the disease. Furthermore, current treatments are limited by the emergence of severe toxicities and multidrug resistance. There is an essential need for economical and relevant research tools to improve our understanding and overcome these problems. This review provides a comprehensive summary of studies that utilized zebrafish for different aims in prostate cancer research. We discuss the use of zebrafish in prostate cancer cell proliferation and metastasis, defining signaling pathways, drug discovery and therapeutic development against prostate cancer, and toxicity studies. Finally, this review highlights limitations in this field and future directions to efficiently use zebrafish as a robust model for prostate cancer therapeutics development.
Collapse
Affiliation(s)
- Haneen Amawi
- Department of Pharmacy Practice, Faculty of Pharmacy, Yarmouk University, P.O.BOX 566, Irbid, 21163, Jordan.
| | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Sai H S Boddu
- College of Pharmacy and Health Sciences, Ajman University, Ajman, UAE
| | - Sadam Amawi
- Department of Urology and General Surgery, Faculty of Medicine, King Abdullah University Hospital, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Mohammad A Obeid
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, St. John's University, Queens, USA
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, The University of Toledo, Toledo, OH, USA.
| |
Collapse
|
45
|
Armando F, Ferrari L, Arcari ML, Azzali G, Dallatana D, Ferrari M, Lombardi G, Zanfabro M, Di Lecce R, Lunghi P, Cameron ER, Cantoni AM, Corradi A. Endocanalicular transendothelial crossing (ETC): A novel intravasation mode used by HEK-EBNA293-VEGF-D cells during the metastatic process in a xenograft model. PLoS One 2020; 15:e0239932. [PMID: 33085676 PMCID: PMC7577447 DOI: 10.1371/journal.pone.0239932] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 09/15/2020] [Indexed: 12/15/2022] Open
Abstract
In cancer metastasis, intravasation of the invasive tumor cell (TCi) represents one of the most relevant events. During the last years, models regarding cancer cell intravasation have been proposed, such as the “endocanalicular transendothelial crossing” (ETC) theory. This theory describes the interplay between two adjacent endothelial cells and the TCi or a leukocyte during intravasation. Two endothelial cells create a channel with their cell membranes, in which the cell fits in without involving endothelial cell intercellular junctions, reaching the lumen through a transendothelial passage. In the present study, ten SCID mice were subcutaneously xenotransplanted with the HEK-EBNA293-VEGF-D cell line and euthanized after 35 days. Post-mortem examinations were performed and proper specimens from tumors were collected. Routine histology and immunohistochemistry for Ki-67, pAKT, pERK, ZEB-1, TWIST-1, F-actin, E-cadherin and LYVE-1 were performed followed by ultrastructural serial sections analysis. A novel experimental approach involving Computed Tomography (CT) combined with 3D digital model reconstruction was employed. The analysis of activated transcription factors supports that tumor cells at the periphery potentially underwent an epithelial-to-mesenchymal transition (EMT)-like process. Topographical analysis of LYVE-1 immunolabeled lymphatics revealed a peritumoral localisation. TEM investigations of the lymphatic vessels combined with 3D digital modelling enhanced the understanding of the endotheliocytes behavior during TCi intravasation, clarifying the ETC theory. Serial ultrastructural analysis performed within tumor periphery revealed numerous cells during the ETC process. Furthermore, this study demonstrates that ETC is an intravasation mode more frequently used by the TCi than by leukocytes during intravasation in the HEK-EBNA293-VEGF-D xenograft model and lays down the potential basis for promising future studies regarding intravasation blocking therapy.
Collapse
Affiliation(s)
- Federico Armando
- Department of Veterinary Science, Pathology Unit, University of Parma, Parma, Italy
- * E-mail: (AMC); (FA); (LF)
| | - Luca Ferrari
- Department of Veterinary Science, Pathology Unit, University of Parma, Parma, Italy
- * E-mail: (AMC); (FA); (LF)
| | | | - Giacomo Azzali
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Davide Dallatana
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Maura Ferrari
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna “B. Ubertini”, Unit of Brescia, Brescia, Italy
| | - Guerino Lombardi
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna “B. Ubertini”, Unit of Brescia, Brescia, Italy
| | - Matteo Zanfabro
- Practitioner, 3D Veterinary Printing Project, Parma, Italy
- Department of Veterinary Science, Diagnostic Imaging Unit, University of Parma, Parma, Italy
| | - Rosanna Di Lecce
- Department of Veterinary Science, Pathology Unit, University of Parma, Parma, Italy
| | - Paolo Lunghi
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma,Parma, Italy
- Centre for Molecular and Translational Oncology, University of Parma, Parma, Italy
| | - Ewan R. Cameron
- School of Veterinary Medicine, University of Glasgow, Glasgow, Scotland
| | - Anna M. Cantoni
- Department of Veterinary Science, Pathology Unit, University of Parma, Parma, Italy
- * E-mail: (AMC); (FA); (LF)
| | - Attilio Corradi
- Department of Veterinary Science, Pathology Unit, University of Parma, Parma, Italy
| |
Collapse
|
46
|
Cabezas-Sáinz P, Pensado-López A, Sáinz B, Sánchez L. Modeling Cancer Using Zebrafish Xenografts: Drawbacks for Mimicking the Human Microenvironment. Cells 2020; 9:E1978. [PMID: 32867288 PMCID: PMC7564051 DOI: 10.3390/cells9091978] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/07/2020] [Accepted: 08/19/2020] [Indexed: 02/07/2023] Open
Abstract
The first steps towards establishing xenografts in zebrafish embryos were performed by Lee et al., 2005 and Haldi et al., 2006, paving the way for studying human cancers using this animal species. Since then, the xenograft technique has been improved in different ways, ranging from optimizing the best temperature for xenografted embryo incubation, testing different sites for injection of human tumor cells, and even developing tools to study how the host interacts with the injected cells. Nonetheless, a standard protocol for performing xenografts has not been adopted across laboratories, and further research on the temperature, microenvironment of the tumor or the cell-host interactions inside of the embryo during xenografting is still needed. As a consequence, current non-uniform conditions could be affecting experimental results in terms of cell proliferation, invasion, or metastasis; or even overestimating the effects of some chemotherapeutic drugs on xenografted cells. In this review, we highlight and raise awareness regarding the different aspects of xenografting that need to be improved in order to mimic, in a more efficient way, the human tumor microenvironment, resulting in more robust and accurate in vivo results.
Collapse
Affiliation(s)
- Pablo Cabezas-Sáinz
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (P.C.-S.); (A.P.-L.)
| | - Alba Pensado-López
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (P.C.-S.); (A.P.-L.)
- Genomic Medicine Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Bruno Sáinz
- Departamento de Bioquímica, Facultad de Medicina, Instituto de Investigaciones Biomédicas “Alberto Sols” CSIC-UAM, Universidad Autónoma de Madrid, Arzobispo Morcillo 4, 28029 Madrid, Spain;
- Cancer Stem Cell and Fibroinflammatory Microenvironment Group, Chronic Diseases and Cancer Area 3-Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (P.C.-S.); (A.P.-L.)
| |
Collapse
|
47
|
Long-term in vivo imaging reveals tumor-specific dissemination and captures host tumor interaction in zebrafish xenografts. Sci Rep 2020; 10:13254. [PMID: 32764590 PMCID: PMC7411039 DOI: 10.1038/s41598-020-69956-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 07/15/2020] [Indexed: 12/15/2022] Open
Abstract
Understanding mechanisms mediating tumor metastasis is crucial for diagnostic and therapeutic targeting. Here, we take advantage of a transparent embryonic zebrafish xenograft model (eZXM) to visualize and track metastatic cells in real time using selective plane illumination microscopy (SPIM) for up to 30 h. Injected human leukemic and breast cancer cells exhibited cell-type specific patterns of intravascular distribution with leukemic cells moving faster than breast cancer cells. Tracking of tumor cells from high-resolution images revealed acute differences in intravascular speed and distance covered by cells. While the majority of injected breast cancer cells predominantly adhered to nearby vasculature, about 30% invaded the non-vascularized tissue, reminiscent of their metastatic phenotype. Survival of the injected tumor cells appeared to be partially inhibited and time-lapse imaging showed a possible role for host macrophages of the recipient embryos. Leukemic cell dissemination could be effectively blocked by pharmacological ROCK1 inhibition using Fasudil. These observations, and the ability to image several embryos simultaneously, support the use of eZXM and SPIM imaging as a functional screening platform to identify compounds that suppress cancer cell spread and invasion.
Collapse
|
48
|
Abstract
Metastasis, the dispersal of cancer cells from a primary tumor to secondary sites within the body, is the leading cause of cancer-related death. Animal models have been an indispensable tool to investigate the complex interactions between the cancer cells and the tumor microenvironment during the metastatic cascade. The zebrafish (Danio rerio) has emerged as a powerful vertebrate model for studying metastatic events in vivo. The zebrafish has many attributes including ex-utero development, which facilitates embryonic manipulation, as well as optically transparent tissues, which enables in vivo imaging of fluorescently labeled cells in real time. Here, we summarize the techniques which have been used to study cancer biology and metastasis in the zebrafish model organism, including genetic manipulation and transgenesis, cell transplantation, live imaging, and high-throughput compound screening. Finally, we discuss studies using the zebrafish, which have complemented and benefited metastasis research.
Collapse
Affiliation(s)
- Katy R Astell
- The Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| | - Dirk Sieger
- The Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| |
Collapse
|
49
|
Cellot G, Vranic S, Shin Y, Worsley R, Rodrigues AF, Bussy C, Casiraghi C, Kostarelos K, McDearmid JR. Graphene oxide nanosheets modulate spinal glutamatergic transmission and modify locomotor behaviour in an in vivo zebrafish model. NANOSCALE HORIZONS 2020; 5:1250-1263. [PMID: 32558850 DOI: 10.1039/c9nh00777f] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Graphene oxide (GO), an oxidised form of graphene, is widely used for biomedical applications, due to its dispersibility in water and simple surface chemistry tunability. In particular, small (less than 500 nm in lateral dimension) and thin (1-3 carbon monolayers) graphene oxide nanosheets (s-GO) have been shown to selectively inhibit glutamatergic transmission in neuronal cultures in vitro and in brain explants obtained from animals injected with the nanomaterial. This raises the exciting prospect that s-GO can be developed as a platform for novel nervous system therapeutics. It has not yet been investigated whether the interference of the nanomaterial with neurotransmission may have a downstream outcome in modulation of behaviour depending specifically on the activation of those synapses. To address this problem we use early stage zebrafish as an in vivo model to study the impact of s-GO on nervous system function. Microinjection of s-GO into the embryonic zebrafish spinal cord selectively reduces the excitatory synaptic transmission of the spinal network, monitored in vivo through patch clamp recordings, without affecting spinal cell survival. This effect is accompanied by a perturbation in the swimming activity of larvae, which is the locomotor behaviour generated by the neuronal network of the spinal cord. Such results indicate that the impact of s-GO on glutamate based neuronal transmission is preserved in vivo and can induce changes in animal behaviour. These findings pave the way for use of s-GO as a modulator of nervous system function.
Collapse
Affiliation(s)
- Giada Cellot
- Department of Neuroscience, Psychology and Behaviour, College of Life Sciences, University of Leicester, Leicester, LE1 7RH, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Frantz WT, Ceol CJ. From Tank to Treatment: Modeling Melanoma in Zebrafish. Cells 2020; 9:cells9051289. [PMID: 32455885 PMCID: PMC7290816 DOI: 10.3390/cells9051289] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/16/2020] [Accepted: 05/18/2020] [Indexed: 12/24/2022] Open
Abstract
Melanoma is the deadliest form of skin cancer and one of few cancers with a growing incidence. A thorough understanding of its pathogenesis is fundamental to developing new strategies to combat mortality and morbidity. Zebrafish—due in large part to their tractable genetics, conserved pathways, and optical properties—have emerged as an excellent system to model melanoma. Zebrafish have been used to study melanoma from a single tumor initiating cell, through metastasis, remission, and finally into relapse. In this review, we examine seminal zebrafish studies that have advanced our understanding of melanoma.
Collapse
Affiliation(s)
- William Tyler Frantz
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA;
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Craig J Ceol
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA;
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
- Correspondence:
| |
Collapse
|